CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017) doi:10.21873/cgp.20041

Review

Histone Deacetylases as New Therapeutic Targets in Triple- negative Breast Cancer: Progress and Promises NIKOLAOS GARMPIS 1* , CHRISTOS DAMASKOS 1,2* , ANNA GARMPI 3* , EMMANOUIL KALAMPOKAS 4, THEODOROS KALAMPOKAS 5, ELEFTHERIOS SPARTALIS 2, AFRODITE DASKALOPOULOU 2, SERENA VALSAMI 6, MICHAEL KONTOS 7, AFRODITI NONNI 8, KONSTANTINOS KONTZOGLOU 1, DESPINA PERREA 2, NIKOLAOS NIKITEAS 2 and DIMITRIOS DIMITROULIS 1

1Second Department of Propedeutic Surgery, Laiko General Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece; 2N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 3Internal Medicine Department, Laiko General Hospital, University of Athens Medical School, Athens, Greece; 4Gynaecological Oncology Department, University of Aberdeen, Aberdeen, U.K.; 5Assisted Conception Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 6Blood Transfusion Department, Aretaieion Hospital, Medical School, National and Kapodistrian Athens University, Athens, Greece; 7First Department of Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 8First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece

Abstract. Triple-negative breast cancer (TNBC) lacks therapies. Given the fact that epigenetic processes control both expression of estrogen receptor (ER), progesterone receptor the initiation and progression of TNBC, there is an increasing (PR) and HER2 gene. It comprises approximately 15-20% of interest in the mechanisms, molecules and signaling pathways breast cancers (BCs). Unfortunately, TNBC’s treatment that participate at the epigenetic modulation of genes continues to be a clinical problem because of its relatively expressed in carcinogenesis. The acetylation of histone poor prognosis, its aggressiveness and the lack of targeted proteins provokes the transcription of genes involved in cell therapies, leaving chemotherapy as the mainstay of treatment. growth, and the expression of histone deacetylases (HDACs) It is essential to find new therapies against TNBC, in order to is frequently up-regulated in many malignancies. surpass the resistance and the invasiveness of already existing Unfortunately, in the field of BC, HDAC inhibitors have shown limited effect as single agents. Nevertheless, their use in combination with kinase inhibitors, autophagy inhibitors, This article is freely accessible online. ionizing radiation, or two HDAC inhibitors together is currently being evaluated. HDAC inhibitors such as *These Authors contributed equally to this study. suberoylanilidehydroxamic acid (SAHA), , , , , YCW1 and N-(2- Correspondence to: Nikolaos Garmpis, MD, Second Department of hydroxyphenyl)-2-propylpentanamide have shown promising Propedeutic Surgery, Laiko General Hospital, National and therapeutic outcomes against TNBC, especially when they are Kapodistrian University of Athens, Medical School, 17 Agiou used in combination with other anticancer agents. More Thoma Street, 11527, Athens, Greece. Tel: +30 6976291403, Fax: +30 2132061772, e-mail: [email protected] studies concerning HDAC inhibitors in breast carcinomas along with a more accurate understanding of the TNBC’s Key Words: , HDAC inhibitors, breast cancer, pathobiology are required for the possible identification of triple negative, epigenetics, review. new therapeutic strategies.

299 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017)

According to the World Health Organization, breast cancer Table I. Classification of breast cancers and their relation to breast (BC) is the most frequently diagnosed cancer and the second cancer cell lines. leading cause of cancer-related death, among women Classification Immuno-profile Cell line worldwide. Interestingly, according to the American Cancer Society about 12% of women in U.S.A. will develop BC Luminal A ER+, PR±, HER2- MCF-7, T47D during their lifetime (1-4). There is a variety of risk factors Luminal B ER+, PR±, HER2+ PR BT-474, ZR-75 that are associated with BC such as gender, age, ethnicity or HER2+ ER –, PR –, HER2+ MDA-MB-453 TNBC (Basal-like) ER –, PR –, HER2 – BT-20, MDA-MB-468 lifestyle, but it is now clear that the most dangerous factor for TNBC (Claudin-low) ER –, PR –, HER2 – BT549, Hs578T, the development of BC is heredity. In particular, in high-risk MDA-MB-231 families, there have been reported more than 1.000 mutations of breast cancer 1 ( BRCA1 ) and breast cancer 2 ( BRCA2 ) TNBC: Triple-negative breast cancer; ER: estrogen receptor; PR: genes (1). progesteron receptor. BC is a very heterogeneous disease given that its invasive process is associated with a variety of molecular alterations. These changes lead to different subtypes of the disease. The HDACs and their Μechanism of Αction current classification divides BC subtypes into Luminal A, Luminal B, HER2-positive and TNBC (5, 6). A better Today, several BC subtypes are treated with many different explanation of BC classification and the corresponding cell primary therapeutic protocols, but none of them uses lines are presented in Table I (7, 8). epigenetic drugs, despite the fact that the interest for First of all, Luminal A and B represent from 65% to 80% of epigenetics in BC tumorgenesis is increasing (12, 13). total BC disease. More specifically they account 50-60% and Gene regulation is affected by nucleosome packaging. 15-20% respectively (9). These subtypes are generally Nucleosome is the organizing DNA structure that is consisted correlated to a good prognosis. They are known to express the of about a 200 bp DNA coiled around an octameric core, estrogen receptor (ER-positive) that participates in cell consisted of histone proteins (14). proliferation, viability, and invasion of BC cells. We should N-terminal tails of histones extend outward from the mention that patients with luminal B tumors frequently present nucleosomal core and are modified by the covalent addition a worse prognosis than Luminal A patients. This could be of groups to the side chains of certain amino acids. These explained as in Luminal B tumors HER2 gene, which is modifications, including methylation, acetylation, associated with potent proliferation, can be amplified (10). The phosphorylation and the reverse processes, can change the substantial difference though, between Luminal A and B secondary DNA structure. As a result, gene promoter regions patients is the cell proliferation rate which is higher in the latter. become accessible or inaccessible to transcription factors (15). Secondly, there is the subtype of HER2-positive patients Histone acetyltransferases (HATs) catalyze the reversible who amplify the oncogene HER2 . These tumors are ER process of lysine acetylation at the ε- amino group of negative, so they are different from Luminal B cancers. proteinogenic lysine residues. Histone acetylation neutralizes Unfortunately, hormonotherapy is inefficient against HER2- the positive charge of lysine residues, therefore is correlated positive tumors. During the last 20 years the development to chromatin relaxation and active gene transcription (17). On of novel drugs targeting HER2 ( e.g. trastuzumab, lapatinib, the other hand, functional antagonists of HATs, histone pertuzumab) has enhanced the clinical outcomes (1). deacetylases (HDACs) remove the acetyl groups (16), thus Moreover, another severe subtype of BC is triple negative leading to compressed chromatin structure (heterochromatin), breast cancer (TNBC). It is associated with poor prognosis. and subsequently suppressing gene transcription (18) (Figure TNBC does not express either estrogen, or progesterone 1). Except for the direct effect of acetylation on chromatin receptors, or HER2 gene. These tumors can be further structure, gene regulation through acetylation is based on classified in several subtypes. The first subgroup is basal-like, synergistic actions. The specific acetylation patterns on histone where tumors express some characteristics of breast tails recruit further chromatin modulators that form co- myoepithelial cells. Basal-like tumors are highly proliferative repressor or co-activator complexes. and are associated with very poor prognosis. Another Several human HDACs have been identified; recently subgroup is claudin-low, which presents epithelial to HDACs have been classified into four classes in accordance mesenchymal transition (EMT) and stem cell-like or/and to functional criteria and homology to yeast proteins (19). In tumor initiating cell features (11). This subtype is also general, HDACs can be divided into Zn 2+ -dependent classes associated with poor prognosis. (class I, II and IV) and NAD-dependent classes (class III). Neoadjuvant anthracycline/taxane-based chemotherapies Class I is consisted of HDACs 1, 2, 3 and 8. Class II can be (e.g. , docetaxel, doxorubicin, cyclophosphamide) have been divided further into class IIa (HDAC4, 5, 7, and 9) and class tested in TNBC patients with poor prognosis (5). IIb (HDAC6 and 10). Class III members, or sirtuins as they

300 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review)

Figure 1. Therapeutic strategy targeting histone deacetylases against triple-negative breast cancer and histone deacetylase inhibitors classification. TNBC: Triple-negative breast cancer; HAT: histone acetyltransferases; HDAC: histone deacetylases.

are often called since they are homologous to silent Clinical trials (phases I and II) have also demonstrated that information regulator 2 (SIR2) of Saccharomyces cerevisiae , HDAC inhibitors result in minor adverse effects in patients consist of SIRT1-SIRT7. Finally, class IV has only one (15, 26-30). Their mechanism of action involves binding of member, HDAC11 (20) (Figure 1). their hydroxamate group to the zinc cation (Zn 2+ ) located in the HDAC cavity (31). Several clinical trials seemed to have HDAC Inhibitors as Anti-cancer Agents a beneficial result. For instance, the US Food and Drug Administration has approved SAHA and as HDAC inhibitors can be classified into four classes according regimen of cutaneous T-cell lymphoma (29, 31) and peripheral to their chemical structure: a) hydroxamates ( e.g. T-cell lymphoma (27) respectively. Moreover, panobinostat suberanilohydroxamic acid (SAHA)), b) ( e.g. MS- treatment is reported to be clinically successful against 275), c) cyclic peptides ( e.g. romidepsin) and d) aliphatic acids multiple myeloma (28). (e.g. valproic acid). Alternatively, HDAC inhibitors can be In total, advanced stages of clinical trials have studied classified according to their specificity for HDAC subtypes or several HDAC inhibitors against many cancer types. However, classes. For example, SAHA and are pan-HDAC concerning the TNBC field, studies have shown that, in inhibitors, while MS-275 and romidepsin inhibit class I and general, HDAC inhibitors succeed clinically beneficial results valproic acid inhibits class I and IIa HDACs (21). as complementary treatment ( e.g. SAHA and VPA), or in It is clear that both histone acetylation and deacetylation combination with cytotoxic drugs and ionizing radiation affect chromatin remodeling as strong epigenetic (Table II). mechanisms. Interestingly, evidence from several reports indicates that HDAC levels are increased in certain cancer SAHA. SAHA or is a pan-HDAC inhibitor that types (22-24). In addition, HDAC inhibitors have been induces apoptosis in several types of haematological and solid reported to enhance the acetylation of histones, in tumor tumor cells (32). Clinical investigations show that SAHA is a cells (25). Unlike other cytostatic-type compounds, HDAC potent inhibitor against tumor types at doses that were well inhibitors have been reported to exert much lower cytoxicity tolerated by patients (33). on normal cells, than on cancer cells. In general, HDAC According to a study in human TNBC cell lines, MDA- inhibitors induce the inhibition of tumor growth and the MB-231 and inBT-549, SAHA significantly promotes in vitro apoptosis of cancer cells. motility via activation of epithelial–mesenchymal transition

301 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017)

Table II. Histone deacetylase inhibitors and their action against triple-negative breast cancer.

HDAC inhibitors Co-treatment Action

SAHA Alone Promotion of EMT via HDAC8/FOXA1 signals Olaparid Inhibition of tumor growth, apoptotic + autophagic cell death IR Inhibition of tumor growth, DNA damage Nucleosome conformation, activation of caspase-3, relegation of beclin-1, inhibition of autophagy Sorafenib Apoptotic + autophagic cell death Cisplatin Inhibition of HDAC6, apoptosis, cell cycle arrest Ferrocifen Anti-proliferative activity NaB Anti-proliferative activity, cell cycle arrest at G 0/G 1 phase, apoptosis, decrease of phosphorylation, protein and mRNA expression of wtp53 NaB TSA G6PDH activation, rising of oxygen consumption Mocetinostat Alone Inhibition of genes involved in cell cycle regulation JQ1 Decrease of cell viability, increase in the expression of USP17L5,decrease of Ras activity Panobinostat Alone Apoptosis, anti-proliferative activity, inhibition of primary tumor volumes, increase of CDH1, cell cycle arrest at G 2/M phase, restoration of ERa mRNA+ protein expression, anti-EMT effects Salinomycin Anti-proliferative activity, inhibition of breast cancer stem cell properties, suppression of ALDH1 activity Chloroquine Inhibition of tumor growth, increase in viability Entinostat Alone Anti-EMT effects, increase of E-cadherin, inhibition of HDAC1, inhibition of cell migration, prevention of metastasis ATRA+Doxorubicin Inhibition of tumor growth, inhibition of Topo II- β, silence of RAR- β YCW1 IR Autophagic cell death, inhibition of BNIP3 Compound 2 Alone Anti-proliferative activity, target HDAC8

HDAC: Histone deacetylases; SAHA: suberoylanilidehydroxamic acid; NaB: sodium butyrate; YCW1: [3-(2-(5-methoxy-1H-indol- 1yl)ethoxy)phenyl]-amide N-hydroxyamide; IR: ionizing radiation; TSA: trichostatin A; ATRA: all-trans retinoic acid; EMT: epithelial to mesenchymal transition; FOXA1; forkhead-box protein A1; G6PDH: glucose-6phosphate dehydrogenase; CDH1: cadherin; ERa: estrogen receptor a; ALDH1: aldehyde dehydrogenase 1; TopoII- β: topoisomerase II- β; RAR- β: retinoic acid receptor; BNIP3: BCL2/adenovirus E1B 19 kDa protein- interacting protein 3.

(EMT) phenotype (34). This phenotype is known as the first silencing of HDAC8, but not HDAC6 (34). Previous studies and critical point for metastatic dissemination of cancer cells. have suggested that HDAC6 and HDAC8 mediate the SAHA Vimentin up-regulation and E-cadherin down-regulation are effects in BC cells (39, 40). HDAC8 lacks the conserved C- considered as markers of EMT (35, 36). terminal domain (41), so it is a unique class I HDAC and it Wu et al. found that SAHA treatment induces the has a different expression profile from those of HDACs 1-3 expression of the mesenchymal markers N-Cadherin, Vimentin (42). HDAC8 silencing does not change histone acetylation and Fibronectin, while decreases the expression of epithelial (43), but it has been assumed that it could act at certain marker E-Cadherin. Nevertheless, SAHA treatment does not promoter sites of FOXA1 . Based on this evidence, researchers change neither the expression nor the nuclear translocation of claimed that HDAC8/FOXA1 signals motivated by SAHA transcription factors related to EMT. These factors were zinc treatment induce the EMT of TNBC cells (34). finger proteins SNAI1 (SNAIL) and SNAI2 (SLUG), basic As it is known, HDACs participate at the homologous helix-loop-helix transcription factor Twist-related protein recombination repair pathway of DNA, given the fact that they (TWIST) and Zinc finger E-box-binding homeobox (ZEB). As regulate the expression of genes related to this procedure (15). a result it seemed that these epithelial–mesenchymal transition Studies have shown that ADP-ribose (poly) polymerase related transcription factors are not involved in this process. (PARP) inhibitors, such as olaparid, could be used as anti- Moreover, SAHA treatment decreased mRNA and protein cancer treatment). Min et al. have searched whether SAHA expression of forkhead-box protein A 1 (FOXA1) (34). treatment enhances the anti-tumor effect of poly (ADP-ribose) FOXA1, as a growth factor, mediates the hormonal response polymerase (PARP) inhibitors in TNBC cells by blocking the in BC (37, 38). When FOXA1 is over-expressed, SAHA homologous recombination repair pathway (46). Investigators induced EMT of TNBC cells is limited. used MDA-MB-157, -231, -453, -468 and BT-549 human cells Furthermore, in MDA-MB-231 and BT-549 cells, SAHA- and found that combinational treatment with olaparid and induced down-regulation of FOXA1 transcription and up- SAHA suppressed the proliferative signaling pathway and regulation of N-cadherin and vimentin were attenuated by promoted inhibition of tumor growth, in comparison to

302 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review) olaparid or SAHA treatment alone. An in vivo study of a terminal tails of histones H3 and H4 and these findings were MDA-MB-231 xenograft model confirmed this evidence. in accordance to other studies (63, 64). Furthermore, Moreover, this combination seemed to induce both apoptotic investigators claimed that there is an association between this and autophagic cell death, thus enhanced the cytotoxic effects histone hyperacetylation provoked by SAHA or SAHA and of these inhibitors (46). Further studies could examine more PN treatment and the up-regulation of tumor suppressor combined treatments of PARP inhibitors with other HDAC factors p21 and p27, as well as down-regulation of inhibitors in order to improve therapeutic approaches for fundamental survival proteins Bcl-2 (55). These results have TNBC patients. also been confirmed by another study (65). This evidence Furthermore, it is known that the majority of deaths in demonstrates that epigenetic actions could induce the TNBC patients are not due to the primary tumors, but to alterations in the expression of these proteins. metastases. An in vitro study on MDA-MB-231 and 4T1 (a Investigators have also studied the production of reactive mouse TNBC cell line) cell lines for the investigation of the oxygen species (ROS) and the autophagic process and they effects of ionizing radiation (IR) and SAHA co-treatment, have found that SAHA treatment urges both processes. Both indicated that the combination was therapeutically more ROS generation and autophagy are reduced by two NADPH effective in a statistically significant way, than IR or SAHA oxidase family (NOX) inhibitors - Nox enzymes are major alone. Tumor growth was also inhibited, in vivo , in an ROS producers-, apocynin and diphenyleneiodonium chloride orthotopic BC mouse model (47). In addition, this combination (DPI). Therefore, it was proposed that SAHA primarily increased DNA damage through the inhibition of DNA repair induced NOX to increase the production of ROS, which led proteins. Concerning SAHA treatment in 4T1 cells, it led to to activation of autophagy (55). Another study has shown that inhibition of BC cell migration and invasion, via the inhibition SAHA stimulates autophagy in tumor cells (66). When they of matrix metalloproteinase-9 (MMP-9) activity. added PN, SAHA’s results on ROS generation and autophagic In general, evidence shows, in several cancer types, MMPs process were diminished, while they observed a potent mediate the invasion and metastasis (48). MMP-9 participates activation of apoptosis. at the degradation of type IV collagen and its over-expression Finally, it has been found that SAHA and PN combined has been found to be associated with invasion and metastatic therapy induced a decrease of the expression levels of beclin-1, potential in many types of carcinomas (49-53). In total, a protein involved in autophagosome formation (55). Previous evidence indicates that SAHA could serve as a radiosensitizer studies have shown that beclin-1 is cleaved by caspases (67). against TNBC (47). N-terminal cleavage fragment of beclin-1 suppresses autophagy, Also, in the same study, an in vivo experimental metastasis while its C-terminal cleavage fragment promotes the release of mouse model was used and showed that SAHA treatment cytochrome C from mitochondria (68). As an extension to the inhibited lung metastasis and enhanced radiosensitivity (47). above mentioned finding, investigators have claimed that Concerning metastases, another study of an in vivo model of SAHA/PN combination treatment relegates beclin-1 by TNBC has reported that SAHA treatment is involved in the activating caspase-3. This event could inhibit SAHA- induced induction of DNA double-strand breaks suppressing brain autophagy and the release of cytochrome C (55). Taken together metastatic colonization by 62% (54). Further studies that this evidence, it could be implied that SAHA/PN co-treatment combine SAHA with DNA active drugs or radiation are could be a candidate for TNBC therapy. needed in order to investigate thoroughly their actions against A recent study on MDA-MB-231 cells used SAHA in metastases. comparison to a kinase inhibitor and found that this co- Carlisi et al. studied the combination of SAHA and treatment induces cell death. An important fact was that cell parthenolide (PN) (55). The principal bioactive sesquiterpene death was induced by both autophagy and apoptosis (69). lactone component of feverfew Tanacetum parthenium , PN, These results were confirmed by Zhang et al. , who used was found to induce tumor inhibition on many cancer types, SAHA and the antiangiogenic kinase inhibitor Sorafenib in such as osteosarcoma, melanoma (56) and prostatic (57), epithelial tumor cell types and found that this treatment pancreatic (58), colorectal (59) cancers. At the same time it suppressed the expression of anti-apoptotic proteins and showed low toxicity against normal cells. PN acts increased the activation of CD95 extrinsic apoptotic and cytotoxically on MDA-MB231 cells, but at low doses it is not lysosomal protease pathways. So this combination has been effective (60, 61). Investigators have combined PN and SAHA found to interact synergistically against liver, kidney, and in order to overcome the PN’s deficiency (55). pancreatic tumor cells in vitro (70). These studies indicate that They claimed that pre-treatment of MDA-MB231 cells with the combination of a kinase and an HDAC inhibitor is a SAHA sensitized the cells to the cytotoxic effect of PN (62), promising treatment modality for the prevention or therapy of so PN and SAHA acted synergistically in order to fight against aggressive cancer types, especially TNBC. TNBC cells (55). Moreover, they demonstrated that SAHA The effects of HDAC inhibitors (VPA and SAHA) and treatment leads to acetylation of lysine residues in the NH 2- cisplatin (CDDP), a chemotherapeutic agent, co-treatment

303 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017) against several types of BC cells have been explored using an protein levels of mutant p53 (mtp53), while neither of these isobolographic method (71,72). This combination achieved to compounds had anti-proliferative effects on wild type p53 reduce the doses of the compounds and to be therapeutically (wtp53) via time and dose dependent manners (84). p53 gene beneficial for head and neck squamous cell carcinomas (26, has been reported as the most frequently mutated gene in BC 73) and BC in vitro (74, 75). This study also showed that in (85). Approximately, 62% of the basal-like TNBC and 43% of MDA-MB231 cells, the combination of the CDDP and VPA the non-basal-like TNBC have mutations in the mtp53 (86). at the ratio of 1:1 induced antagonistic interactions, while These data urged the investigators to report that the CDDP and SAHA co-treatment induced synergistic inhibition occurs during the transcription, given that the interactions (71). According to the investigators, this could be transcription of the precursor p53 is down-regulated more explained by the fact that SAHA is a potent inhibitor of the rapidly (less than 2h) and aggressively than that of mature HDAC6 activity compared to VPA (26). Many nuclear p53. Also, the transcription of mtp53 is inhibited by the proteins, like the transcription factor forkhead boxp3 (FOXP3) silencing of HDAC8, while not of HDAC6 (84). Both and the DNA repair factor KU70, are substrates of HDAC6, HDAC6 and HDAC8 are previously reported to mediate the which is recruited by RNA polymerase II to chromatin in down-regulation of p53 induced by HDAC inhibitors (40, 87). order to suppress the cycles of acetylation/deacetylation. This Moreover, both SAHA and NaB reduced the binding of procedure enables the transcription and transfers cytotoxic transcription factor Yin Yang 1 (YY1) with the -102 to -96 poly-ubiquitinylated proteins into autophagosomes (76). All position of human p53 promoter. Interestingly, silencing of these mechanisms of HDAC6 action and its potential YY1 significantly inhibits the mtp53 transcription (83). YY1 inhibition by SAHA could explain the hypothesis. is suggested to be over-expressed in BC cells and to be a key Moreover, it has been reported that CDDP/HDAC inhibitor molecule in BC progression. Knockdown of YY1 led to co-treatment induces apoptosis and cell-cycle arrest of TNBC suppression of the clonogenicity, migration, invasion and cells (71). Further research in vivo is necessary in order to use tumor formation of BC cells (88). this promising combination therapy in TNBC clinical trials. Based on the above, researchers have found that both A recent study has described a new family of hybrid SAHA and NaB reduced the association of HDAC8 and YY1. compounds that combine particular structural motifs of As a result, acetylation of residues 170–200 of YY1 was tamoxifen (TAM), ferrocifen (FcTAM) and SAHA (77). enhanced, leading to decreased YY1 transcription and finally, Ferrocenyl group (Fc) is reported to act as a redox antenna for inhibition of the YY1- induced p53 transcription (84). In total, phenol oxidation via an intra-molecular mechanism (78-80). this study showed that not only SAHA and NaB treatment They have found that ferrocene derivatives are extremely could be very promising for TNBC patients, but also more active than organic analogs in MDA-MB-231 cells and HDAC8/YY1/mtp53 signals could become an important target hormone-dependent MCF-7 BC cell lines. FcTAM improved for TNBC therapy. its inhibitory activity by replacing its 3-(dimethylamino) propan-1oxy group with an 8-hydroxyamino-8-oxooctanamido Trichostatin A and NaB. Trichostatin A (TSA) is an antifungal or an 8-amino-8-oxooctanamido group. In MDA-MB-231 BC antibiotic, found in cultured mammalian cells and, at low cells, FcTAM–SAHA treatment exerted better anti- nanomolar concentrations, in fractionated cell nuclear extracts. proliferative effects than SAHA treatment alone (77). In general, as an HDAC inhibitor, TSA not only suppresses Interestingly, the organometallic compounds induced the HDAC activity, but also leads to cell-cycle arrest in G 1 and expression of p21waf1/cip1 gene in MCF-7 cells in G2 phase, induces cell differentiation as well as the reversion accordance with their antiproliferative activity (77). Further of transformed cells in vitro (89). research on the field of organometallic moiety could A study of the effect of NaB and TSA on viability of non- contribute to a more proper treatment against TNBC. tumorgenic MCF10A cells and of MCF-7, T-47D and MDA- MB-231 cell lines (90) has found that MCF10A cells were Sodium Butyrate and SAHA. Sodium butyrate (NaB) is shown minimum affected by elevated doses of these HDAC to exhibit beneficial anticancer ability (81). It has the potential inhibitors while the other tested cell lines were extremely to relax the chromatin structure, allowing easier access to sensitive to treatment with both NaB and TSA. Concerning transcription-related proteins. These abilities render NaB a the TNBC cell line (MDA-MB-231), NaB treatment potent treatment against several types of solid tumor (82, 83). significantly enhanced the activity of pyruvate kinase, but it A study in MDA-MB-231 and BT-549 cell lines using various neither induced an attenuation of glycolysis, nor enhanced the concentrations of NaB and SAHA for 24 h revealed that these activity of lactate dehydrogenase, as it happened in T-47 D inhibitors suppress cell proliferation, arrest cell cycle at G 0/G 1 cells. Furthermore, in both T-47D and MDA-MB-231 cells, phase, and finally are involved in mitochondrial-related the rate-limiting enzyme of the pentose phosphate pathway, apoptosis. Moreover, both SAHA and NaB treatment Glucose-6phosphate dehydrogenase (G6PDH) was activated decreased the phosphorylation, and also the mRNA and by NaB treatment, while in MCF-7 cells, G6PDH activity was

304 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review) inhibited. Finally, both NaB and TSA in MDA-MB-231 and in hematologic, lung, breast, ovarian, thyroid and prostate T-47D cells significantly raised the oxygen consumption. malignancies (104). Recently panobinostat was approved for These data show that mitochondria might occupy a key role treatment of multiple myeloma with tumor progression after in metastasis (90). Further research is required in order to immunomodulating agents and bortezomib (105). understand the pathways of action of these inhibitors and then Interestingly, a study has reported significant action of target the proper signals to suppress the specific BC type. panobinostat against the TNBC, as it exerted anti-proliferative action, provoked apoptosis and inhibited the primary tumor Mocetinostat. Mocetinostat is a class I HDAC inhibitor that is volumes of TNBC xenografts (106). Also in the same study, associated with the reversion of cardiac fibrosis (91). Recently, in vitro and in vivo results have shown that treatment with mocetinostat was reported to have promising antitumor panobinostat increases significantly the epithelial cell marker activities against various types of cancer cell lines and tumor CDH1. In general, the expression of EMT markers, such as xenografts in nude mice, as it is related to apoptosis (92-94). A epithelial-cadherin (CDH1), neuronal-cadherin (CDH2), study showed that mocetinostat inhibits the growth of colon vimentin (VIM), zinc finger E-box binding homeobox 1 cancer cells via the up-regulation of WNT ligand DKK-1 (ZEB1) and 2 (ZEB2), has been associated with poor expression (95). Also, in B-cell chronic lymphocytic leukemia prognosis of BC patients (107-109). As a result, these markers cells, it has been shown to lead to apoptosis, as it decreases the have become attractive targets for the confrontation of expression of Mcl-1 protein and it induces Bax translocation to metastasis. mitochondria (96). Another study has shown that mocetinostat In a recent study it has been tested whether panobinostat has inhibits the proliferation of prostate cancer cells (93). the ability to inhibit the migration, invasion, and metastasis of Borbely et al. studied the combination of mocetinostat and MDA-MB-231 and BT-549 cell lines, by suppressing EMT JQ1 in MDA-MB-231, BT549, MCF-7 and T47-D cells (97). (110). In TNBC cells, panobinostat has been reported to inhibit JQ1 is a bromodomain and extra-C terminal (BET) inhibitor the expression of genes associated to EMT and to induce the and is reported to participate in the expression of oncogenes expression of CDH1, while these results were not found in and tumor suppressor genes. JQ1 is in pre-clinical testing for luminal A or basal BC cell lines (106). This study has also the treatment of hematological malignancies and reported very important results about panobinostat effects on neuroblastoma (98-100). Researchers found that either JQ1 or MDA-MB-231 and BT-549 cells, as it augmented histone mocetinostat treatment alone, inhibited the expression of genes acetylation, while suppressed the cell proliferation and viability. involved in cell cycle regulation. Interestingly, the It led to cell cycle arrest at G 2/M phase with a simultaneous combination of JQ1 and mocetinostat decreased even more decrease in S phase. Moreover, panobinostat induced apoptosis cell viability as the compounds acted synergistically. at 24 h. In addition, treatment of ER-negative breast cancer Furthermore, combination treatment was associated with a cells (MDA-MB-231 and MDA-MB-435) with panobinostat sharp increase in the expression of many members of the for 24 h resulted in restoration of ER α mRNA and protein ubiquitin–specific protease 17 (USP17) family of expression. However, reactivation of ER α expression was not deubiquitinating enzymes (97). Previous studies have reported due to changes in the methylation profile of CpG island within that USP17 regulates Ras/MAPK signaling partly through the ER promoter region. Interestingly, after at least 96 h from regulation of the RCE1 (101). It is known that the Ras/MAPK termination of panobinostat treatment, the expression of ER α pathway is involved in BC progression (102). The activity of this mRNA was maintained. Panobinostat treatment released DNA pathway has been tested and it was found that the expression of (cytosine-5)-methyltransferase 1 (DNMT1), HDAC1, and the USP17L5 protein was increased, when at the same time Ras H3 lysine 9 (H3-K9) methyltransferase SUV39H1 from the ER activity was decreased. In other words, USP17 enzymes promoter. Investigators claimed that these changes altered the attenuated the Ras/MAPK pathway and cell viability was chromatin state to active, characterized by increased the levels suppressed. On the other hand siRNA-mediated depletion of of acetylated H3 and H4, decreased methylated H3-K9 and USP17, decreased the cytoxicity, indicating that the Ras/MAPK false binding of heterochromatin protein 1 (HP1 alpha) at the signaling pathway is involved in the synergistic effect of the promoter (111). combinational treatment (97). All these data show that the Furthermore, Rhodes et al. compared panobinostat to other combination of HDAC and BET inhibitors could be used as HDAC inhibitors (SAHA, TMP269) and found that potential therapeutic strategy for BC, given that it reduces the panobinostat has much more potent anti-EMT effects than the viability of TNBC cells, through induction of USP17. other inhibitors (110). In vitro research showed that changes in EMT-related gene expression were associated with the Panobinostat. Panobinostat (LBH589) is a pan-HDAC suppression of cell migration and invasion. Also, in a inhibitor. It has the ability to block several pathways related xenograft model these alterations have induced a statistically to cancer and reverse epigenetic events related to cancer significant inhibition of metastasis of TNBC cells to both progression (103). Its anti-cancer effects have been observed brain and lung. Except for CDH1expression, panobinostat has

305 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017) induced the suppression of multiple predominant pro-EMT A study on MDA-MB-231cells investigated the in vitro and genes including ZEB1 and ZEB2 . In addition, when in vivo effect of co-treatment of panobinostat with the investigators forced exogenous expression of ZEB1 or ZEB2 , autophagy inhibitor chloroquine (123). First of all, the inhibitory effects of panobinostat on cell migration, panobinostat treatment disrupted the hsp90/HDAC6/HSF1/p97 invasion, and CDH1 expression were reversed (110). complex (124, 125) and induced a heat shock response It has been recently reported that ZEBs recruit Class I mediated by heat shock factor protein 1 (HSF1). This HDACs (48, 112, 113). Given the fact that panobinostat can abruption of the HSF1 from the multiprotein comlex is act against Class I, II and IV HDACs, in addition to inhibition reported to lead to HSF1 phosphorylation, trimerization and of ZEB expression, panobinostat has the ability to prevent nuclear translocation (125, 126). Moreover, panobinostat ZEB-mediated repression of CDH1. treatment induced endoplasmic reticulum stress response and However, it is probably the repression of ZEB2 that autophagy (123), evidence also supported by previous research regulates the effects of panobinostat on tumor genesis, as (127-129). Combinational treatment of panobinostat with ZEB1 expression alone was not sufficient to overcome the chloroquine was found to significantly enhance both the in actions of panobinostat on CDH1 expression (110). To sum vivo and the in vitro effect of panobinostat, given the fact that up, it is critical to study panobinostat as a novel therapeutic to it inhibited tumor growth and increased viability of MDA- target the aggressive and metastatic nature of TNBC, as MB-231 xenografts (123). panobinostat is claimed to inhibit both TNBC primary tumor Therefore, panobinostat could be used for treatment of genesis and the metastasis of TNBC cells. aggressive BC that is resistant to hormonal therapy. We Another study has confirmed these findings, given that it propose that panobinostat should be tested either in was found that in MDA-MB231 cells independent of estrogen monotherapy, or in combination with known anti-cancer receptor expression, panobinostat induced CDH1 expression agents, like trastuzumab, capecitabine, lapatinib, or paclitaxel. (114). Panobinostat was reported to induce the expression of CDH1 and reduce the migration and invasion of TNBC cells. Entinostat. Previous studies have reported that retinoic acid Researchers claimed that panobinostat by-passed the CDH1 and its products, such as all-trans retinoic acid (ATRA), urge transcriptional repressors (Snail, Slug), while the ERa the differentiation of multiple types of stem cells, including expression and pathway were not affected. the stem cells that exist in BC (130, 131). On the other hand, Another combination that has been studied on TNBC cell ATRA has been used in a clinical trial and its efficacy was lines (HCC1937 and MDA-MB-231) is co-treatment of limited (132), possibly because of the epigenetic knock-out of panobinostat and salinomycin (115). Salinomycin is an the retinoic acid receptor (RAR)- β (133). Recent studies have antibiotic that has been used in farm animals (116) and it has shown that HDAC inhibitors have the ability to re-express been associated with the inhibition of BC stem cells in TNBC RAR- β, so they restore the sensitivity of the cells to treatment (117, 118). Also, studies in glioblastoma have shown that co- (134-136). It has been reported that co-treatment of HDAC treatment of salomycin and HDAC inhibitors induced the inhibitors with either doxorubicin or retinoids increased the death of stem-like glioblastoma cells (119). Kai et al. have cytoxicity against cancer cells (75, 137). reported that panobinostat and salinomycin acted A novel study has examined the effects of co-treatment with synergistically and inhibited the TNBC cell proliferation (115). entinostat (MS-275), which is a selective inhibitor of class I HDAC inhibitors have been found to be beneficial in BC HDACs, ATRA, and doxorubicin and reported that TNBC cell therapy, as they target BC stem cells, have anti-proliferative growth was inhibited in xenografts and killed tumor cells and effects on aldehyde dehydrogenase 1 (ALDH1)-positive cells, in cell cultures (138). Given the fact that direct binding of and suppress the formation of BC mammospheres (120). Also, topoisomerase II- β ( TopoII- β) to RAR- β promoter has been ALDH1 has been associated with TNBC, so ALDH1 can be reported in acute pro-myelocytic leukemia cells (139), used as a BC stem cell marker in TNBC (121, 122). Based on investigators have searched that path and found that entinostat these facts, Kai et al. determined the activity of panobinostat and doxorubicin treatment suppressed the expression of Topo on TNBC cells with BC stem cell properties (115). Co- II- β. As a result, RAR- β expression was silenced. It was treatment of panobinostat and salinomycin inhibited the BC demonstrated that this co-treatment achieved significant tumor stem cell properties, as the self-renewal capacity and ALDH1 suppression in TNBC, as entinostat urges the cytoxicity activity were suppressed. Moreover, this combinational mediated by doxorubicin and the differentiation mediated by treatment inhibited growth of ALDH1-positive cells, induced retinoid (138). apoptosis, arrested cell cycle and regulated EMT in BC stem As we mentioned, in metastatic cell lines and invasive BCs cells (115). We can assume that further research is needed, E-cadherin is silenced epigenetically and EMT phenotype is given that co-treatment of panobinostat with salinomycin in reported to be the critical step in the beginning of metastasis TNBC is claimed to provide an effective targeted therapy for (140-142). In MDA-MB-231 and Hs578T cells, entinostat patients with TNBC. treatment has been reported to reverse the EMT phenotype

306 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review)

(143). These cell lines have lost the expression of E-cadherin, the most promising results between other derivatives that were while they highly express mesenchymal markers such as N- submitted to docking simulations. In vitro studies against cadherin and vimentin along with transcriptional repressors HeLa, rhabdomyosarcoma and BC cells lines have shown that such as Twist and Snail (144-147). Treatment with entinostat compound 2 was the best anticancer agent (154). Interestingly, increased the transcription of E-cadherin, while the mRNA it was effective against TNBC cells. It seemed that compound expression of N-cadherin was decreased. Chromatin 2 targeted HDAC8. In addition, in vitro studies have shown immunoprecipitation analysis demonstrated that E-cadherin’s that compound 2 inhibited cancer cell proliferation at a much promoter was activated due to a decrease in the association of lower concentration, compared to valproic acid (154). TWIST and SNAIL with E-cadherin promoter (143). In a previous study, it has been reported that HDAC1 activity is Uncommon Results required for the SNAIL-mediated repression of E-cadherin (145), so investigators studied the activity of HDAC1 and Unfortunately, despite the promising results in several pre- found that entinostat inhibits HDAC1, thus the repression of clinical trials, when HDAC inhibitors are used as single E-cadherin is reversed. An in vitro study has shown that agents, they failed to be clinically beneficial against solid entinostat inhibited cell migration. Moreover, there was tumors (33). For instance, in the field of metastatic BC, SAHA observed an increase of vimentin phosphorylation levels, as treatment in 14 patients failed to achieve adequate single-agent well as changes in vimentin filaments. Interestingly, the activity (155). Moreover, de Cremoux et al. have used increased phosphorylation of vimentin led to suppression of different HDAC inhibitors (SAHA, panobinostat, ) the formation of microtentacles based on tubulin (143), which in MDA-MB231, Hs 578T and SUM149 human cell lines and are known to help floating cells attach to other surfaces (148). found that despite the cytotoxicity of both of these inhibitors We should also mention that a subset of cells within a Estrogen receptor 1 (ESR1) and estrogen receptor 2 (ESR2) breast tumor may cause escape from the primary site. These genes were not re-expressed in vitro (156). Xenograft studies cells are named as tumor-initiating cells (TICs). TICs produce with abexinostat treatment for three consecutive days did not progenitor cells, which cannot self-renew, but they may induce the expression of ESR1/ESR1 related genes or of ERa comprise the greater part of the tumor (149). Entinostat protein. In addition, intra-tumor H3 acetylation was observed treatment on TNBC cells has been shown to decrease the level (156). Another study in MDA-MB231 and SKBR3 lines of TICs, therefore entinostat might be used to prevent treated with SAHA or valproic acid for 48 h, showed minimal development of metastasis (150). or no alterations in ERa or in ERb mRNa and protein (157). It is apparent that the ability of entinostat to reverse the Also, different HDAC inhibitors have been shown to cause EMT phenotype and reduce the migration of TNBC cells, as different effects on luminal and mesenchymal-like breast it reduces the attachment of floating cancer cells to new cancer cells, either elevating histone acetylation or showing surfaces, render it a promising therapeutic agent against no effects (158). metastasis. Conclusion YCW1. The octanedioic acid [3-(2-(5-methoxy-1 H-indol- 1yl)ethoxy)phenyl]-amide N-hydroxyamide (YCW1) is a TNBC is a serious subtype of BC that is correlated with poor novel HDAC inhibitor that has been developped by structure- prognosis due to the high proliferation rate. New therapeutic based analyses (151, 152). Researchers have tested the co- strategies are needed in order to cure TNBC. Epigenetic treatment with YCW1 and ionizing radiation (IR) in a murine mutations are associated with repression of BC development and a human TNBC cell line, 4 T1, and MDA-MB-231, and therapy resistance. respectively. This combination was shown to increase Between all the epigenetic treatments, HDAC inhibitors autophagy and endoplasmic reticulum stress, so these represent the first successful anti-cancer epigenetic therapy. compounds acted as a cytotoxic. Interistingly, in comparison Studies show that they affect positively the therapy of to the effects of SAHA, YCW1 significantly enhanced subtypes of hematological malignancies. However, it is still toxicity. Study in an orthotopic BC mouse model confirmed unclear how effective they are against solid tumors (159). these results (153). Therefore, co-treatment with IR and Nevertheless, the majority of studies in the field of TNBC YCW1 induced autophagic cell death. therapy tend to combine HDAC inhibitors with kinase inhibitors, autophagy inhibitors, antibiotics, chemotherapy, IR, N-(2-Hydroxyphenyl)-2propylpentanamide. N -(2-Hydroxyphe- or even two HDAC inhibitors treatment in order to enhance nyl)-2propylpentanamide (Compound 2) is an aryl derivative their efficacy against TNBC (47, 69, 71, 84, 115, 123, 153). of valproic acid. These derivatives are produced by combining In the majority of the studies, co-treatment of an HDAC valproic acid and the arylamine core of SAHA with different inhibitor with another compound induced the inhibition of substituents at its carboxyl group. Compound 2 has exerted tumor growth and showed anti-proliferative effects (46, 47,

307 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017)

77, 84, 115, 123, 138, 154). Also, several HDAC inhibitors Lippman M, Ethier S, Gazdar A and Gray JW: A collection of are assosiated not only with autophagic cell death, but also breast cancer cell lines for the study of functionally distinct apoptosis (44, 55, 70, 71, 84, 97, 153). Interestingly, many cancer subtypes. Cancer Cell 10 : 515-527, 2006. 9 Bernhardt SM, Dasari P, Walsh D, Townsend AR, Price TJ and studies have found that HDAC inhibitors have the ability to Ingman WV: Hormonal Modulation of Breast Cancer Gene convert ER-negative tumors to ER-positive tumors (111, 160, Expression: Implications for Intrinsic Subtyping in 161). Finally, a recent study showed that entinostat treatment Premenopausal Women. Front Oncol 6: 241, 2016. can prevent the metastasis (143). 10 Mohamed A, Krajewski K, Cakar B and Ma CX: Targeted However, as we mentioned above, we should not forget that therapy for breast cancer. Am J Pathol 183 : 1096-1112, 2013. there are several studies in the pertinent literature with 11 Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz contradictory results (155-157). These unexpected studies JI, He X and Perou CM: Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast reveal the necessity for further investigation in the field of cancer. Breast Cancer Res 12 : R68, 2010. HDAC inhibitors against TNBC cancer. We should understand 12 Marks DL, Olson RL and Fernandez-Zapico ME: Epigenetic the nature of the molecular basis of the selectivity of HDAC control of the tumor microenvironment. Epigenomics 8: 671- inhibitors and which are the proper combining treatments in 1687, 2016. order to find more clinically beneficial treatment against 13 Jones P: The cancer epigenome. Genome 56 : 540-541, 2013. TNBC. 14 Rando OJ and Ahmad K: Rules and regulation in the primary structure of chromatin. Curr Opin Cell Biol 19 : 250-256, 2007. Conflicts of Interest 15 Woo YM: Epigenetic regulation in cystogenesis. Adv Exp Med Biol 933: 59-68, 2016. The Authors declare that they do not have any conflict of interest. 16 Kuo MH and Allis CD: Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 20 : 615-626, 1998. Acknowledgements 17 Kouzarides T: Chromatin modifications and their function. Cell 128 : 693-705, 2007. 18 Haberland M, Montgomery RL and Olson EN: The many roles of Christos Damaskos would like to express his gratitude to his teacher, histone deacetylases in development and physiology: implications Spyridon Garmpis who not only selflessly taught him, but also his for disease and therapy. Nat Rev Genet 10 : 32-42, 2009. actions inspired him to learn more, do more and become better. 19 Trapp J and Jung M: The role of NAD+ dependent histone deacetylases (sirtuins) in ageing. Curr Drug Targets 7: 1553- References 1560, 2006. 20 Olzscha H, Sheikh S and La Thangue NB: Deacetylation of 1 Antoniou AC and Easton DF: Models of genetic susceptibility to chromatin and gene expression regulation: a new target for breast cancer. Oncogene 25 : 5898-5905, 2006. epigenetic therapy. Crit Rev Oncog 20 : 1-17, 2015. 2 Rivenbark AG, O’Connor SM and Coleman WB: Molecular and 21 Marks PA: The clinical development of histone deacetylase cellular heterogeneity in breast cancer: challenges for inhibitors as targeted anticancer drugs. Expert Opin Investig personalized medicine. Am J Pathol 183 : 1113-1124, 2013. Drugs 19 : 1049-1066, 2010. 3 Nyante SJ, Lee SS, Benefield TS, Hoots TN and Henderson LM: 22 Zhang Z, Yamashita H, Toyama T, Sugiura H, Ando Y, Mita K, The association between mammographic calcifications and breast Hamaguchi M, Hara Y, Kobayashi S and Iwase H: Quantitation cancer prognostic factors in a population-based registry cohort. of HDAC1 mRNA expression in invasive carcinoma of the Cancer 123 : 219-227, 2017. breast. Breast Cancer Res Treat 94 : 11-16, 2005. 4 DeSantis CE, Bray F, Ferlay J, Lortet-Tieulent J, Anderson BO 23 Stojanovic N, Hassan Z, Wirth M, Wenzel P, Beyer M, Schäfer and Jemal A: International variation in female breast cancer C, Brand P, Kroemer A, Stauber RH, Schmid RM, Arlt A, incidence and mortality rates. Cancer Epidemiol Biomarkers Prev Sellmer A, Mahboobi S, Rad R, Reichert M, Saur D, Krämer OH 24 : 1495-1506, 2015. and Schneider G: HDAC1 and HDAC2 integrate the expression 5 Claude-Taupin A, Boyer-Guittaut M, Delage-Mourroux R and of p53 mutants in pancreatic cancer. Oncogene 36 : 1804-1815, Hervouet E: Use of epigenetic modulators as a powerful adjuvant 2017. for breast cancer therapies. Methods Mol Biol 1238 : 487-509, 24 Li A, Liu Z, Li M, Zhou S, Xu Y, Xiao Y and Yang W: HDAC5, 2015. a potential therapeutic target and prognostic biomarker, promotes 6 Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees proliferation, invasion and migration in human breast cancer. CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Oncotarget 7: 37966-37978, 2016. Pergamenschikov A, Williams C, Zhu SX, Lønning PE, 25 New M, Olzscha H and La Thangue NB: HDAC inhibitor-based Børresen-Dale AL, Brown PO and Botstein D: Molecular therapies: can we interpret the code? Mol Oncol 6: 637-656, portraits of human breast tumours. Nature 406 : 747-752, 2000. 2012. 7 Holliday DL and Speirs V: Choosing the right cell line for breast 26 Lee JH, Choy ML, Ngo L, Foster SS and Marks PA: Histone cancer research. Breast Cancer Res 13 : 215, 2011. deacetylase inhibitor induces DNA damage, which normal but 8 Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark not transformed cells can repair. Proc Natl Acad Sci USA 107 : L, Bayani N, Coppe JP, Tong F, Speed T, Spellman PT, DeVries 14639-14644, 2010. S, Lapuk A, Wang NJ, Kuo WL, Stilwell JL, Pinkel D, Albertson 27 Barbarotta L and Hurley K: Romidepsin for the treatment of DG, Waldman FM, McCormick F, Dickson RB, Johnson MD, peripheral T-cell lymphoma. J Adv Pract Oncol 6: 22-36, 2015.

308 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review)

28 Libby EN, Becker PS, Burwick N, Green DJ, Holmberg L and 44 Gunderson CC and Moore KN: Olaparib: an oral PARP-1 and Bensinger WI: Panobinostat: a review of trial results and future PARP-2 inhibitor with promising activity in ovarian cancer. prospects in multiple myeloma. Expert Rev Hematol 8: 9-18, Future Oncol 11 : 747-757, 2015. 2015. 45 Arun B, Akar U, Gutierrez-Barrera AM, Hortobagyi GN and 29 Mann BS, Johnson JR, He K, Sridhara R, Abraham S, Booth BP, Ozpolat B: The PARP inhibitor AZD2281 (Olaparib) induces Verbois L, Morse DE, Jee JM, Pope S, Harapanhalli RS, Dagher autophagy/mitophagy in BRCA1 and BRCA2 mutant breast R, Farrell A, Justice R and Pazdur R: Vorinostat for treatment of cancer cells. Int J Oncol 47 : 262-268, 2015. cutaneous manifestations of advanced primary cutaneous T-cell 46 Min A, Im SA, Kim DK, Song SH, Kim HJ, Lee KH, Kim TY, lymphoma. Clin Cancer Res 13 : 2318-2322, 2007. Han SW, Oh DY, Kim TY, O’Connor MJ and Bang YJ: Histone 30 Damaskos C, Garmpis N, Valsami S, Spartalis E, Antoniou EA, deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), Tomos P, Karamaroudis S, Zoumpou T, Pergialiotis V, Stergios enhances anti-tumor effects of the poly (ADP-ribose) polymerase K, Michaelides C, Kontzoglou K, Perrea D, Nikiteas N and (PARP) inhibitor olaparib in triple-negative breast cancer cells. Dimitroulis D: Histone deacetylase inhibitors: A Novel Breast Cancer Res 17 : 33, 2015. therapeutic weapon against medullary thyroid cancer? Anticancer 47 Chiu HW, Yeh YL, Wang YC, Huang WJ, Chen YA, Chiou YS, Res 36 : 5019-5024, 2016. Ho SY, Lin P and Wang YJ: Suberoylanilide hydroxamic acid, 31 Marks PA: Discovery and development of SAHA as an an inhibitor of histone deacetylase, enhances radiosensitivity and anticancer agent. Oncogene 26 : 1351-1356, 2007. suppresses lung metastasis in breast cancer in vitro and in vivo . 32 Marks P, Rifkind RA, Richon VM, Breslow R, Miller T and PLoS One 8: e76340, 2013. Kelly WK: Histone deacetylases and cancer: causes and 48 Wang J, Xu H, Wang Q, Zhang H, Lin Y, Zhang H, Li Q and therapies. Nat Rev Cancer 1: 194-202, 2001. Panq T: CIAPIN1 targets Na(+)/H(+) exchanger 1 to mediate 33 Slingerland M, Guchelaar HJ and Gelderblom H: Histone MDA-MB-231 cells’ metastasis through regulation of MMPs via deacetylase inhibitors: an overview of the clinical studies in solid ERK1/2 signaling pathway. Exp Cell Res 333 : 60-72, 2015. tumors. Anticancer Drugs 25 : 140-149, 2014. 49 Cupić DF, Tesar EC, Ilijas KM, Nemrava J and Kovacević M: 34 Wu S, Luo Z, Yu PJ, Xie H and He YW: Suberoylanilide Expression of matrix metalloproteinase 9 in primary and hydroxamic acid (SAHA) promotes the epithelial mesenchymal recurrent breast carcinomas. Coll Antropol 35(Suppl 2) : 7-10, transition of triple negative breast cancer cells via 2011. HDAC8/FOXA1 signals. Biol Chem 397 : 75-83, 2016. 50 Li Y, Wu T, Zhang B, Yao Y and Yin G: Matrix 35 Zavadil J and Bottinger EP: TGF-beta and epithelial-to- metalloproteinase-9 is a prognostic marker for patients with mesenchymal transitions. Oncogene 24 : 5764-5774, 2005. cervical cancer. Med Oncol 29 : 3394-3399, 2012. 36 Zeisberg M and Neilson EG: Biomarkers for epithelial- 51 Hu X, Li D, Zhang W, Zhou J, Tang B and Li L: Matrix mesenchymal transitions. J Clin Invest 119 : 1429-1437, 2009. metalloproteinase-9 expression correlates with prognosis and 37 Wolf I, Bose S, Williamson EA, Miller CW, Karlan BY and involved in ovarian cancer cell invasion. Arch Gynecol Obstet Koeffler HP: FOXA1: Growth inhibitor and a favorable 286 : 1537-1543, 2012. prognostic factor in human breast cancer. Int J Cancer 120 : 1013- 52 Tang FY, Chiang EP and Sun YC: Resveratrol inhibits heregulin- 1022, 2007. beta1-mediated matrix metalloproteinase-9 expression and cell 38 Robinson JL and Carroll JS: FoxA1 is a key mediator of invasion in human breast cancer cells. J Nutr Biochem 19 : 287- hormonal response in breast and prostate cancer. Front 294, 2008. Endocrinol (Lausanne) 3: 68, 2012. 53 Tang D, Piao Y, Zhao S, Mu X, Li S, Ma W, Song Y, Wang J, 39 Kim YJ, Greer CB, Cecchini KR, Harris LN, Tuck DP and Kim Zhao W and Zhang Q: Expression and correlation of matrix TH: HDAC inhibitors induce transcriptional repression of high metalloproteinase-9 and heparanase in patients with breast copy number genes in breast cancer through elongation blockade. cancer. Med Oncol 31 : 26, 2014. Oncogene 32 : 2828-2835, 2013. 54 Palmieri D, Lockman PR, Thomas FC, Hua E, Herring J, 40 Yan W, Liu S, Xu E, Zhang J, Zhang Y, Chen X and Chen X: Hargrave E, Johnson M, Flores N, Qian Y, Vega-Valle E, Taskar Histone deacetylase inhibitors suppress mutant p53 transcription KS, Rudraraju V, Mittapalli RK, Gaasch JA, Bohn KA, via histone deacetylase 8. Oncogene 32 : 599-609, 2013. Thorsheim HR, Liewehr DJ, Davis S, Reilly JF, Walker R, 41 Somoza JR, Skene RJ, Katz BA, Mol C, Ho JD, Jennings AJ, Bronder JL, Feigenbaum L, Steinberg SM, Camphausen K, Luong C, Arvai A, Buggy JJ, Chi E, Tang J, Sang BC, Verner E, Meltzer PS, Richon VM, Smith QR and Steeg PS: Vorinostat Wynands R, Leahy EM, Dougan DR, Snell G, Navre M, Knuth inhibits brain metastatic colonization in a model of triple- MW, Swanson RV, McRee DE and Tari LW: Structural snapshots negative breast cancer and induces DNA double-strand breaks. of human HDAC8 provide insights into the class I histone Clin Cancer Res 15 : 6148-6157, 2009. deacetylases. Structure 12 : 1325-1334, 2004. 55 Carlisi D, Lauricella M, D’Anneo A, Buttitta G, Emanuele S, 42 Hu E, Chen Z, Fredrickson T, Zhu Y, Kirkpatrick R, Zhang GF, di Fiore R, Martinez R, Rolfo C, Vento R and Tesoriere G: Johanson K, Sung CM, Liu R and Winkler J: Cloning and The synergistic effect of SAHA and parthenolide in MDA- characterization of a novel human class I histone deacetylase that MB231 breast cancer cells. J Cell Physiol 230 : 1276-1289, functions as a transcription repressor. J Biol Chem 275 : 15254- 2015. 15264, 2000. 56 D’Anneo A, Carlisi D, Lauricella M, Emanuele S, Di Fiore R, 43 Oehme I, Deubzer HE, Wegener D, Pickert D, Linke JP, Hero B, Vento R and Tesoriere G: Parthenolide induces caspase- Kopp-Schneider A, Westermann F, Ulrich SM, von Deimling A, independent and AIF-mediated cell death in human Fischer M and Witt O: Histone deacetylase 8 in neuroblastoma osteosarcoma and melanoma cells. J Cell Physiol 228 : 952-967, tumorigenesis. Clin Cancer Res 15 : 91-99, 2009. 2013.

309 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017)

57 Sun Y, St Clair DK, Xu Y, Crooks PA and St Clair WH: A Grant S and Dent P: Vorinostat and sorafenib synergistically kill NADPH oxidase-dependent redox signaling pathway mediates tumor cells via FLIP suppression and CD95 activation. Clin the selective radiosensitization effect of parthenolide in prostate Cancer Res 14 : 5385-5399, 2008. cancer cells. Cancer Res 70 : 2880-2890, 2010. 71 Wawruszak A, Luszczki JJ, Grabarska A, Gumbarewicz E, 58 Liu JW, Cai MX, Xin Y, Wu QS, Ma J, Yang P, Xie HY and Dmoszynska-Graniczka M, Polberg K et al : Assessment of Huang DS: Parthenolide induces proliferation inhibition and interactions between cisplatin and two histone deacetylase apoptosis of pancreatic cancer cells in vitro . J Exp Clin Cancer inhibitors in MCF7, T47D and MDA-MB-231 human breast Res 29 : 108, 2010. cancer cell lines - An Isobolographic Analysis. PLoS One 10 : 59 Zhang S, Ong CN and Shen HM: Critical roles of intracellular e0143013, 2015. thiols and calcium in parthenolide-induced apoptosis in human 72 Dasari S and Tchounwou PB: Cisplatin in cancer therapy: colorectal cancer cells. Cancer Lett 208 : 143-153, 2004. molecular mechanisms of action. Eur J Pharmacol 740 : 364-378, 60 D’Anneo A, Carlisi D, Lauricella M, Puleio R, Martinez R, Di 2014. Bella S, Di Marco P, Emanuele S, Di Fiore R, Guercio A, Vento 73 Lin CT, Lai HC, Lee HY, Lin WH, Chang CC, Chu TY, Lin YW, R and Tesoriere G: Parthenolide generates reactive oxygen Lee KD and Yu MH: Valproic acid resensitizes cisplatin-resistant species and autophagy in MDA-MB231 cells. A soluble ovarian cancer cells. Cancer Sci 99 : 1218-1226, 2008. parthenolide analogue inhibits tumour growth and metastasis in 74 Suzuki M, Endo M, Shinohara F, Echigo S and Rikiishi H: a xenograft model of breast cancer. Cell Death Dis 4: e891, 2013. Enhancement of cisplatin cytotoxicity by SAHA involves 61 Carlisi D, D’Anneo A, Martinez R, Emanuele S, Buttitta G, Di endoplasmic reticulum stress-mediated apoptosis in oral Fiore R, Vento R, Tesoriere G and Lauricella M: The oxygen squamous cell carcinoma cells. Cancer Chemother Pharmacol 64 : radicals involved in the toxicity induced by parthenolide in 1115-1122, 2009. MDA-MB-231 cells. Oncol Rep 32 : 167-172, 2014. 75 Kim MS, Blake M, Baek JH, Kohlhagen G, Pommier Y and 62 Lauricella M, Ciraolo A, Carlisi D, Vento R and Tesoriere G: Carrier F: Inhibition of histone deacetylase increases cytotoxicity SAHA/TRAIL combination induces detachment and anoikis of to anticancer drugs targeting DNA. Cancer Res 63 : 7291-7300, MDA-MB231 and MCF-7 breast cancer cells. Biochimie 94 : 2003. 287-299, 2012. 76 Kramer OH, Mahboobi S and Sellmer A: Drugging the HDAC6- 63 Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, HSP90 interplay in malignant cells. Trends Pharmacol Sci 35 : Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi 501-519, 2014. N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, 77 Cázares Marinero Jde J, Lapierre M, Cavaillès V, Saint-Fort R, Marks PA and Anderson KC: Transcriptional signature of histone Vessières A, Top S and Jaouen G: Efficient new constructs deacetylase inhibition in multiple myeloma: biological and against triple negative breast cancer cells: synthesis and clinical implications. Proc Natl Acad Sci USA 101 : 540-545, preliminary biological study of ferrocifen-SAHA hybrids and 2004. related species. Dalton Trans 42 : 15489-15501, 2013. 64 Chen S, Zhao Y, Gou WF, Zhao S, Takano Y and Zheng HC: The 78 Hagen H, Marzenell P, Jentzsch E, Wenz F, Veldwijk MR and anti-tumor effects and molecular mechanisms of suberoylanilide Mokhir A: Aminoferrocene-based prodrugs activated by reactive hydroxamic acid (SAHA) on the aggressive phenotypes of oxygen species. J Med Chem 55 : 924-934, 2012. ovarian carcinoma cells. PLoS One 8: e79781, 2013. 79 Hillard E, Vessières A, Thouin L, Jaouen G and Amatore C: 65 Bali P, Pranpat M, Swaby R, Fiskus W, Yamaguchi H, Balasis Ferrocene-mediated proton-coupled electron transfer in a series M, Rocha K, Wang HG, Richon V and Bhalla K: Activity of of ferrocifen-type breast-cancer drug candidates. Angew Chem suberoylanilide hydroxamic Acid against human breast cancer Int Ed Engl 45 : 285-290, 2005. cells with amplification of her-2. Clin Cancer Res 11 : 6382-6389, 80 Hillard EA, de Abreu FC, Ferreira DC, Jaouen G, Goulart MO 2005. and Amatore C: Electrochemical parameters and techniques in 66 Gammoh N, Lam D, Puente C, Ganley I, Marks PA and Jiang X: drug development, with an emphasis on quinones and related Role of autophagy in histone deacetylase inhibitor-induced compounds. Chem Commun (Camb) 23 : 2612-2628, 2008. apoptotic and nonapoptotic cell death. Proc Natl Acad Sci USA 81 Xiong F, Mou YZ and Xiang XY: Inhibition of mouse B16 109 : 6561-6565, 2012. melanoma by sodium butyrate correlated to tumor associated 67 Gordy C and He YW: The crosstalk between autophagy and macrophages differentiation suppression. Int J Clin Exp Med 8: apoptosis: where does this lead? Protein Cell 3: 17-27, 2012. 4170-4174, 2015. 68 Wirawan E, Vande Walle L, Kersse K, Cornelis S, Claerhout S, 82 Demary K, Wong L and Spanjaard RA: Effects of retinoic acid Vanoverberghe I, Roelandt R, De Rycke R, Verspurten J, and sodium butyrate on gene expression, histone acetylation and Declercq W, Agostinis P, Vanden Berghe T, Lippens S and inhibition of proliferation of melanoma cells. Cancer Lett 163 : Vandenabeele P: Caspase-mediated cleavage of Beclin-1 103-107, 2001. inactivates Beclin-1-induced autophagy and enhances apoptosis 83 Jazirehi AR: Regulation of apoptosis-associated genes by histone by promoting the release of proapoptotic factors from deacetylase inhibitors: implications in cancer therapy. Anticancer mitochondria. Cell Death Dis 1: e18, 2010. Drugs 21 : 805-813, 2010. 69 Librizzi M, Spencer J and Luparello C: Biological effect of a 84 Wang ZT, Chen ZJ, Jiang GM, Wu YM, Liu T, Yi YM, Zeng J, hybrid anticancer agent based on kinase and histone deacetylase Du J and Wang HS: Histone deacetylase inhibitors suppress inhibitors on triple-negative (MDA-MB231) breast cancer cells. mutant p53 transcription via HDAC8/YY1 signals in triple Int J Mol Sci 17 : 1235, 2016. negative breast cancer cells. Cell Signal 28 : 506-515, 2016. 70 Zhang G, Park MA, Mitchell C, Hamed H, Rahmani M, Martin 85 Freed-Pastor WA and Prives C: Mutant p53: one name, many AP, Curiel DT, Yacoub A, Graf M, Lee R, Roberts JD, Fisher PB, proteins. Genes Dev 26 : 1268-1286, 2012.

310 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review)

86 Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili 97 Borbely G, Haldosen LA, Dahlman-Wright K and Zhao C: G, Ding J, Tse K, Haffari G, Bashashati A, Prentice LM, Khattra Induction of USP17 by combining BET and HDAC inhibitors in J, Burleigh A, Yap D, Bernard V, McPherson A, Shumansky K, breast cancer cells. Oncotarget 6: 33623-33635, 2015. Crisan A, Giuliany R, Heravi-Moussavi A, Rosner J, Lai D, Birol 98 Fiskus W, Sharma S, Qi J, Valenta JA, Schaub LJ, Shah B, Peth I, Varhol R, Tam A, Dhalla N, Zeng T, Ma K, Chan SK, Griffith K, Portier BP, Rodriguez M, Devaraj SG, Zhan M, Sheng J, Iyer M, Moradian A, Cheng SW, Morin GB, Watson P, Gelmon K, SP, Bradner JE and Bhalla KN: Highly active combination of Chia S, Chin SF, Curtis C, Rueda OM, Pharoah PD, Damaraju S, BRD4 antagonist and histone deacetylase inhibitor against human Mackey J, Hoon K, Harkins T, Tadigotla V, Sigaroudinia M, acute myelogenous leukemia cells. Mol Cancer Ther 13 : 1142- Gascard P, Tlsty T, Costello JF, Meyer IM, Eaves CJ, Wasserman 1154, 2014. WW, Jones S, Huntsman D, Hirst M, Caldas C, Marra MA and 99 Henssen A, Thor T, Odersky A, Heukamp L, El-Hindy N, Aparicio S: The clonal and mutational evolution spectrum of Beckers A, Speleman F, Althoff K, Schäfers S, Schramm A, Sure primary triple-negative breast cancers. Nature 486 : 395-399, 2012. U, Fleischhack G, Eggert A and Schulte JH: BET bromodomain 87 Li D, Marchenko ND and Moll UM: SAHA shows preferential protein inhibition is a therapeutic option for medulloblastoma. cytotoxicity in mutant p53 cancer cells by destabilizing mutant Oncotarget 4: 2080-2095, 2013. p53 through inhibition of the HDAC6-Hsp90 chaperone axis. 100 Molenaar JJ, Domingo-Fernández R, Ebus ME, Lindner S, Cell Death Differ 18 : 1904-1913, 2011. Koster J, Drabek K, Mestdagh P, van Sluis P, Valentijn LJ, van 88 Wan M, Huang W, Kute TE, Miller LD, Zhang Q, Hatcher H, Nes J, Broekmans M, Haneveld F, Volckmann R, Bray I, Wang J, Stovall DB, Russell GB, Cao PD, Deng Z, Wang W, Heukamp L, Sprüssel A, Thor T, Kieckbusch K, Klein-Hitpass Zhang Q, Lei M, Torti SV, Akman SA and Sui G: Yin Yang 1 L, Fischer M, Vandesompele J, Schramm A, van Noesel MM, plays an essential role in breast cancer and negatively regulates Varesio L, Speleman F, Eggert A, Stallings RL, Caron HN, p27. Am J Pathol 180 : 2120-2133, 2012. Versteeg R and Schulte JH: LIN28B induces neuroblastoma and 89 Yoshida M, Horinouchi S and Beppu T: Trichostatin A and enhances MYCN levels via let-7 suppression. Nat Genet 44 : trapoxin: novel chemical probes for the role of histone acetylation 1199-1206, 2012. in chromatin structure and function. Bioessays 17 : 423-430, 1995. 101 Burrows JF, Kelvin AA, McFarlane C, Burden RE, McGrattan 90 Rodrigues MF, Carvalho É, Pezzuto P, Rumjanek FD and Amoêdo MJ, De la Vega M, Govender U, Quinn DJ, Dib K, Gadina M, ND: Reciprocal modulation of histone deacetylase inhibitors Scott CJ and Johnston JA: USP17 regulates Ras activation and sodium butyrate and trichostatin A on the energy metabolism of cell proliferation by blocking RCE1 activity. J Biol Chem 284 : breast cancer cells. J Cell Biochem 116 : 797-808, 2015. 9587-9595, 2009. 91 Nural-Guvener HF, Zakharova L, Nimlos J, Popovic S, 102 Giltnane JM and Balko JM: Rationale for targeting the Mastroeni D and Gaballa MA: HDAC class I inhibitor, Ras/MAPK pathway in triple-negative breast cancer. Discov Med Mocetinostat, reverses cardiac fibrosis in heart failure and 17 : 275-283, 2014. diminishes CD90+ cardiac myofibroblast activation. Fibrogenesis 103 Atadja P: Development of the pan-DAC inhibitor panobinostat Tissue Repair 7: 10, 2014. (LBH589): successes and challenges. Cancer Lett 280 : 233-241, 92 Zhang Q, Sun M, Zhou S and Guo B: Class I HDAC inhibitor 2009. mocetinostat induces apoptosis by activation of miR-31 104 Ganai SA: Panobinostat: The small molecule metalloenzyme expression and suppression of E2F6. Cell Death Discov 2: inhibitor with marvelous anticancer activity. Curr Top Med Chem 16036, 2016. 16 : 427-434, 2016. 93 Fournel M, Bonfils C, Hou Y, Yan PT, Trachy-Bourget MC, 105 Mu S, Kuroda Y, Shibayama H, Hino M, Tajima T, Corrado C, Kalita A, Liu J, Lu AH, Zhou NZ, Robert MF, Gillespie J, Wang Lin R, Waldron E, Binlich F and Suzuki K: Panobinostat PK/PD JJ, Ste-Croix H, Rahil J, Lefebvre S, Moradei O, Delorme D, profile in combination with bortezomib and dexamethasone in Macleod AR, Besterman JM and Li Z: MGCD0103, a novel patients with relapsed and relapsed/refractory multiple myeloma. isotype-selective histone deacetylase inhibitor, has broad Eur J Clin Pharmacol 72 : 153-161, 2016. spectrum antitumor activity in vitro and in vivo . Mol Cancer Ther 106 Tate CR, Rhodes LV, Segar HC, Driver JL, Pounder FN, Burow 7: 759-768, 2008. ME and Collins-Burow BM: Targeting triple-negative breast 94 Bonfils C, Kalita A, Dubay M, Siu LL, Carducci MA, Reid G, cancer cells with the histone deacetylase inhibitor panobinostat. Martell RE, Besterman JM and Li Z: Evaluation of the Breast Cancer Res 14 : R79, 2012. pharmacodynamic effects of MGCD0103 from preclinical 107 Kong D, Li Y, Wang Z and Sarkar FH: Cancer Stem Cells and models to human using a novel HDAC enzyme assay. Clin Epithelial-to-Mesenchymal Transition (EMT)-Phenotypic Cancer Res 14 : 3441-3449, 2008. Cells: Are They Cousins or Twins? Cancers (Basel) 3: 716- 95 Sikandar S, Dizon D, Shen X, Li Z, Besterman J and Lipkin SM: 729, 2011. The class I HDAC inhibitor MGCD0103 induces cell cycle arrest 108 Eger A, Aigner K, Sonderegger S, Dampier B, Oehler S, and apoptosis in colon cancer initiating cells by upregulating Schreiber M, Berx G, Cano A, Beug H and Foisner R: DeltaEF1 Dickkopf-1 and non-canonical Wnt signaling. Oncotarget 1: 596- is a transcriptional repressor of E-cadherin and regulates 605, 2010. epithelial plasticity in breast cancer cells. Oncogene 24 : 2375- 96 El-Khoury V, Moussay E, Janji B, Palissot V, Aouali N, Brons 2385, 2005. NH, Van Moer K, Pierson S, Van Dyck E and Berchem G: The 109 Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven histone deacetylase inhibitor MGCD0103 induces apoptosis in L, Bruyneel E, Mareel M, Huylebroeck D and van Roy F: The B-cell chronic lymphocytic leukemia cells through a two-handed E box binding zinc finger protein SIP1 mitochondria-mediated caspase activation cascade. Mol Cancer downregulates E-cadherin and induces invasion. Mol Cell 7: Ther 9: 1349-1360, 2010. 1267-1278, 2001.

311 CANCER GENOMICS & PROTEOMICS 14 : 299-313 (2017)

110 Rhodes LV, Tate CR, Segar HC, Burks HE, Phamduy TB, Hoang 124 Vabulas RM, Raychaudhuri S, Hayer-Hartl M and Hartl FU: V, Elliott S, Gilliam D, Pounder FN, Anbalagan M, Chrisey DB, Protein folding in the cytoplasm and the heat shock response. Rowan BG, Burow ME and Collins-Burow BM: Suppression of Cold Spring Harb Perspect Biol 2: a004390, 2010. triple-negative breast cancer metastasis by pan-DAC inhibitor 125 Trepel J, Mollapour M, Giaccone G and Neckers L: Targeting the panobinostat via inhibition of ZEB family of EMT master dynamic HSP90 complex in cancer. Nat Rev Cancer 10 : 537- regulators. Breast Cancer Res Treat 145 : 593-604, 2014. 549, 2010. 111 Zhou Q, Atadja P and Davidson NE: Histone deacetylase 126 Akerfelt M, Morimoto RI and Sistonen L: Heat shock factors: inhibitor LBH589 reactivates silenced estrogen receptor alpha integrators of cell stress, development and lifespan. Nat Rev Mol (ER) gene expression without loss of DNA hypermethylation. Cell Biol 11 : 545-555, 2010. Cancer Biol Ther 6: 64-69, 2007. 127 Rao R, Nalluri S, Fiskus W, Savoie A, Buckley KM, Ha K, 112 Aghdassi A, Sendler M, Guenther A, Mayerle J, Behn CO, Balusu R, Joshi A, Coothankandaswamy V, Tao J, Sotomayor E, Heidecke CD, Friess H, Büchler M, Evert M, Lerch MM and Atadja P and Bhalla KN: Role of CAAT/enhancer binding protein Weiss FU: Recruitment of histone deacetylases HDAC1 and homologous protein in panobinostat-mediated potentiation of HDAC2 by the transcriptional repressor ZEB1 downregulates E- bortezomib-induced lethal endoplasmic reticulum stress in mantle cadherin expression in pancreatic cancer. Gut 61 : 439-448, 2012. cell lymphoma cells. Clin Cancer Res 16 : 4742-4754, 2010. 113 Kakihana M, Ohira T, Chan D, Webster RB, Kato H, Drabkin 128 Tabas I and Ron D: Integrating the mechanisms of apoptosis HA and Gemmill RM: Induction of E-cadherin in lung cancer induced by endoplasmic reticulum stress. Nat Cell Biol 13 : 184- and interaction with growth suppression by histone deacetylase 190, 2011. inhibition. J Thorac Oncol 4: 1455-1465, 2009. 129 Rao R, Nalluri S, Kolhe R, Yang Y, Fiskus W, Chen J, Ha K, 114 Fortunati N, Marano F, Bandino A, Frairia R, Catalano MG and Buckley KM, Balusu R, Coothankandaswamy V, Joshi A, Atadja Boccuzzi G: The pan-histone deacetylase inhibitor LBH589 P and Bhalla KN: Treatment with panobinostat induces glucose- (panobinostat) alters the invasive breast cancer cell phenotype. regulated protein 78 acetylation and endoplasmic reticulum stress Int J Oncol 44 : 700-708, 2014. in breast cancer cells. Mol Cancer Ther 9: 942-952, 2010. 115 Kai M, Kanaya N, Wu SV, Mendez C, Nguyen D, Luu T and 130 Ginestier C, Wicinski J, Cervera N, Monville F, Finetti P, Chen S: Targeting breast cancer stem cells in triple-negative Bertucci F, Wicha MS, Birnbaum D and Charafe-Jauffret E: breast cancer using a combination of LBH589 and salinomycin. Retinoid signaling regulates breast cancer stem cell Breast Cancer Res Treat 151 : 281-294, 2015. differentiation. Cell Cycle 8: 3297-3302, 2009. 116 Miyazaki Y, Shibuya M, Sugawara H, Kawaguchi O and Hirsoe 131 Bhat-Nakshatri P, Goswami CP, Badve S, Sledge GW Jr and C: Salinomycin, a new polyether antibiotic. J Antibiot (Tokyo) Nakshatri H: Identification of FDA-approved drugs targeting 27 : 814-821, 1974. breast cancer stem cells along with biomarkers of sensitivity. Sci 117 Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg Rep 3: 2530, 2013. RA and Lander ES: Identification of selective inhibitors of cancer 132 Connolly RM, Nguyen NK and Sukumar S: Molecular pathways: stem cells by high-throughput screening. Cell 138 : 645-659, 2009. current role and future directions of the retinoic acid pathway in 118 King TD, Suto MJ and Li Y: The Wnt/beta-catenin signaling cancer prevention and treatment. Clin Cancer Res 19 : 1651-1659, pathway: a potential therapeutic target in the treatment of triple 2013. negative breast cancer. J Cell Biochem 113 : 13-18, 2012. 133 Tang XH and Gudas LJ: Retinoids, retinoic acid receptors, and 119 Booth L, Roberts JL, Conley A, Cruickshanks N, Ridder T, Grant cancer. Annu Rev Pathol 6: 345-364, 2011. S, Poklepovic A and Dent P: HDAC inhibitors enhance the 134 Sirchia SM, Ferguson AT, Sironi E, Subramanyan S, Orlandi lethality of low dose salinomycin in parental and stem-like GBM R, Sukumar S and Sacchi N: Evidence of epigenetic changes cells. Cancer Biol Ther 15 : 305-316, 2014. affecting the chromatin state of the retinoic acid receptor beta2 120 Salvador MA, Wicinski J, Cabaud O, Toiron Y, Finetti P, Josselin promoter in breast cancer cells. Oncogene 19 : 1556-1563, E, Lelièvre H, Kraus-Berthier L, Depil S, Bertucci F, Collette Y, 2000. Birnbaum D, Charafe-Jauffret E and Ginestier C: The histone 135 Nacht M, Ferguson AT, Zhang W, Petroziello JM, Cook BP, Gao deacetylase inhibitor abexinostat induces cancer stem cells YH, Maguire S, Riley D, Coppola G, Landes GM, Madden SL differentiation in breast cancer with low Xist expression. Clin and Sukumar S: Combining serial analysis of gene expression Cancer Res 19 : 6520-6531, 2013. and array technologies to identify genes differentially expressed 121 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ and in breast cancer. Cancer Res 59 : 5464-5470, 1999. Clarke MF: Prospective identification of tumorigenic breast 136 Sirchia SM, Ren M, Pili R, Sironi E, Somenzi G, Ghidoni R, cancer cells. Proc Natl Acad Sci USA 100 : 3983-3988, 2003. Toma S, Nicolò G and Sacchi N: Endogenous reactivation of the 122 Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, RARbeta2 tumor suppressor gene epigenetically silenced in Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, Schott A, Hayes breast cancer. Cancer Res 62 : 2455-2461, 2002. D, Birnbaum D, Wicha MS and Dontu G: ALDH1 is a marker of 137 Pili R, Salumbides B, Zhao M, Altiok S, Qian D, Zwiebel J, normal and malignant human mammary stem cells and a predictor Carducci MA and Rudek MA: Phase I study of the histone of poor clinical outcome. Cell Stem Cell 1: 555-567, 2007. deacetylase inhibitor entinostat in combination with 13-cis 123 Rao R, Balusu R, Fiskus W, Mudunuru U, Venkannagari S, retinoic acid in patients with solid tumours. Br J Cancer 106 : 77- Chauhan L, Smith JE, Hembruff SL, Ha K, Atadja P and Bhalla 84, 2012. KN: Combination of pan-histone deacetylase inhibitor and 138 Merino VF, Nguyen N, Jin K, Sadik H, Cho S, Korangath P, Han autophagy inhibitor exerts superior efficacy against triple- L, Foster YM, Zhou XC, Zhang Z, Connolly RM, Stearns V, Ali negative human breast cancer cells. Mol Cancer Ther 11 : 973- SZ, Adams C, Chen Q, Pan D, Huso DL, Ordentlich P, Brodie A 983, 2012. and Sukumar S: Combined treatment with epigenetic,

312 Garmpis et al : Histone Deacetylase Inhibitors in Triple Negative Breast Cancer (Review)

differentiating, and chemotherapeutic agents cooperatively targets 153 Chiu HW, Yeh YL, Wang YC, Huang WJ, Ho SY, Lin P and tumor-initiating cells in triple-negative breast cancer. Cancer Res Wang YJ: Combination of the novel histone deacetylase inhibitor 76 : 2013-2024, 2016. YCW1 and radiation induces autophagic cell death through the 139 McNamara S, Wang H, Hanna N and Miller WH Jr: downregulation of BNIP3 in triple-negative breast cancer cells Topoisomerase IIbeta negatively modulates retinoic acid receptor in vitro and in an orthotopic mouse model. Mol Cancer 15 : 46, alpha function: a novel mechanism of retinoic acid resistance. 2016. Mol Cell Biol 28 : 2066-2077, 2008. 154 Prestegui-Martel B, Bermúdez-Lugo JA, Chávez-Blanco A, 140 van Horssen R, Hollestelle A, Rens JA, Eggermont AM, Schutte Dueñas-González A, García-Sánchez JR, Pérez-González OA, M and Ten Hagen TL: E-cadherin promotor methylation and Padilla-Martínez II, Fragoso-Vázquez MJ, Mendieta-Wejebe JE, mutation are inversely related to motility capacity of breast Correa-Basurto AM, Méndez-Luna D, Trujillo-Ferrara J and cancer cells. Breast Cancer Res Treat 136 : 365-377, 2012. Correa-Basurto J: N-(2-hydroxyphenyl)-2-propylpentanamide, a 141 Guarino M: Epithelial-mesenchymal transition and tumour valproic acid aryl derivative designed in silico with improved invasion. Int J Biochem Cell Biol 39 : 2153-2160, 2007. anti-proliferative activity in HeLa, rhabdomyosarcoma and breast 142 Guarino M, Rubino B and Ballabio G: The role of epithelial- cancer cells. J Enzyme Inhib Med Chem 31(Suppl 3) : 140-149, mesenchymal transition in cancer pathology. Pathology 39 : 305- 2016. 318, 2007. 155. Luu TH, Morgan RJ, Leong L, Lim D, McNamara M, Portnow 143 Shah P, Gau Y and Sabnis G: Histone deacetylase inhibitor J, Frankel P, Smith DD, Doroshow JH, Wong C, Aparicio A, entinostat reverses epithelial to mesenchymal transition of breast Gandara DR and Somlo G: A phase II trial of vorinostat cancer cells by reversing the repression of E-cadherin. Breast (suberoylanilide hydroxamic acid) in metastatic breast cancer: a Cancer Res Treat 143 : 99-111, 2014. California Cancer Consortium study. Clin Cancer Res 14 : 7138- 144 Fearon ER: Connecting estrogen receptor function, 7142, 2008. transcriptional repression, and E-cadherin expression in breast 156. de Cremoux P, Dalvai M, N’Doye O, Moutahir F, Rolland G, cancer. Cancer Cell 3: 307-310, 2003. Chouchane-Mlik O, Assayag F, Lehmann-Che J, Kraus-Berthie 145 Peinado H, Ballestar E, Esteller M and Cano A: Snail mediates L, Nicolas A, Lockhart BP, Marangoni E, de Thé H, Depil S, E-cadherin repression by the recruitment of the Sin3A/histone Bystricky K and Decaudin D: HDAC inhibition does not induce deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol 4: 306- estrogen receptor in human triple-negative breast cancer cell lines 319, 2004. and patient-derived xenografts. Breast Cancer Res Treat 149 : 81- 146 Vesuna F, van Diest P, Chen JH and Raman V: Twist is a 89, 2015. transcriptional repressor of E-cadherin gene expression in breast 157. Biçaku E, Marchion DC, Schmitt ML and Münster PN: cancer. Biochem Biophys Res Commun 367 : 235-241, 2008. Selective inhibition of histone deacetylase 2 silences 147 Baranwal S and Alahari SK: Molecular mechanisms controlling progesterone receptor-mediated signaling. Cancer Res 68 : 1513- E-cadherin expression in breast cancer. Biochem Biophys Res 1519, 2008. Commun 384 : 6-11, 2009. 158. Dagdemir A, Judes G, Lebert A, Echegut M, Karsli-Ceppioglu 148 Whipple RA, Balzer EM, Cho EH, Matrone MA, Yoon JR and S, Rifaï K, Daures M, Ngollo M, Dubois L, Penault-Llorca F, Martin SS: Vimentin filaments support extension of tubulin- Bignon YJ and Bernard-Gallon D: Epigenetic modifications with based microtentacles in detached breast tumor cells. Cancer Res DZNep, NaBu and SAHA in luminal and mesenchymal-like 68 : 5678-5688, 2008. breast cancer subtype cells. Cancer Genomics Proteomics 13 : 149 Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, 291-303, 2016. Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, 159. Rodríguez-Paredes M and Esteller M: Cancer epigenetics Polyak K, Brisken C, Yang J and Weinberg RA: The epithelial- reaches mainstream oncology. Nat Med 17 : 330-339, 2011. mesenchymal transition generates cells with properties of stem 160 Hodges-Gallagher L, Valentine CD, Bader SE and Kushner PJ: cells. Cell 133 : 704-715, 2008. Inhibition of histone deacetylase enhances the anti-proliferative 150 Schech A, Kazi A, Yu S, Shah P and Sabnis G: Histone deacetylase action of antiestrogens on breast cancer cells and blocks inhibitor Entinostat inhibits tumor-initiating cells in triple-negative tamoxifen-induced proliferation of uterine cells. Breast Cancer breast cancer cells. Mol Cancer Ther 14 : 1848-1857, 2015. Res Treat 105 : 297-309, 2007. 151 Huang WJ, Chen CC, Chao SW, Yu CC, Yang CY, Guh JH, Lin 161 Sabnis GJ, Goloubeva O, Chumsri S, Nguyen N, Sukumar S and YC, Kuo CI, Yang P and Chang CI: Synthesis and evaluation of Brodie AM: Functional activation of the estrogen receptor- α and aliphatic-chain hydroxamates capped with osthole derivatives as aromatase by the HDAC inhibitor entinostat sensitizes ER- histone deacetylase inhibitors. Eur J Med Chem 46 : 4042-4049, negative tumors to letrozole. Cancer Res 71 : 1893-1903, 2011. 2011. 152 Huang WJ, Tang YA, Chen MY, Wang YJ, Hu FH, Wang TW, Chao SW, Chiu HW, Yeh YL, Chang HY, Juan HF, Lin P and Wang YC8: A histone deacetylase inhibitor YCW1 with antitumor and antimetastasis properties enhances cisplatin Received May 26, 2017 activity against non-small cell lung cancer in preclinical studies. Revised July 17, 2017 Cancer Lett 346 : 84-93, 2014. Accepted July 19, 2017

313